You are on page 1of 8

Seminars in Cancer Biology 69 (2021) 391–398

Contents lists available at ScienceDirect

Seminars in Cancer Biology


journal homepage: www.elsevier.com/locate/semcancer

Targeted delivery of miRNA based therapeuticals in the clinical


management of Glioblastoma Multiforme
Faheem Hyder Pottoo a, *, Md. Noushad Javed b, c, **, Jawad Ur Rahman d, Tareq Abu-Izneid e,
Firdos Alam Khan f, *
a
Department of Pharmacology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam 31441, Saudi Arabia
b
Department of Pharmaceutics, School of Pharmaceutical Education and Research (SPER), Jamia Hamdard, New-Delhi, India
c
School of Pharmaceutical Sciences, Apeejay Stya University, Gurugram, Haryana, India
d
Department of Microbiology, College of Medicine, Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam, 31441, Saudi Arabia
e
Pharmaceutical Sciences, College of Pharmacy, Al Ain University, Al Ain, Abu Dhabi, United Arab Emirates
f
Department of Stem Cell Research, Institute for Research and Medical consultations (IRMC), Imam Abdulrahman Bin Faisal University, P. O. Box 1982, Dammam,
31441, Saudi Arabia

A R T I C L E I N F O A B S T R A C T

Keywords: Glioblastoma multiforme (GBM) is the most aggressive (WHO grade IV) form of diffuse glioma endowed with
Glioblastoma tremendous invasive capacity. The availability of narrow therapeutic choices for GBM management adds to the
miRNA irony, even the post-treatment median survival time is roughly around 14-16 months. Gene mutations seem to be
nanoparticles
cardinal to GBM formation, owing to involvement of amplified and mutated receptor tyrosine kinase (RTK)-
signaling pathways
BBB
encoding genes, leading to dysregulation of growth factor signaling pathways. Of-late, the role of different
BBTB microRNAs (miRNAs) in progression and proliferation of GBM was realized, which lead to their burgeon po­
nanomedicine tential applications for diagnostic and therapeutic purposes. miRNA signatures are intricately linked with onset
receptor tyrosine kinase and progression of GBM. Although, progression of GBM causes significant changes in the BBB to form BBTB, but
still efficient passage of cancer therapeutics, including antibodies and miRNAs are prevented, leading to low
bioavailability. Recent developments in the nanomedicine field provide novel approaches to manage GBM via
efficient and brain targeted delivery of miRNAs either alone or as part of cytotoxic pharmaceutical composition,
thereby modulating cell signaling in well predicted manner to promise positive therapeutic outcomes.

1. Introduction schwannomas, meningiomas and medulloblastomas. Glioma can be


further classified on the basis of location, and characteristic features of
Glioma is the commonest destructive form of primary brain tumors differentiation or anaplasia. Based on the presence or absence of
affecting 3.19/100000 person in USA alone. Glioma is more prevalent in anaplasia the malignant glioma is classified into subtype I-IV according
caucasians than afro-american, african-asian and american –Indians, to WHO standards [1]. In brief; WHO reclassified the tumors of CNS and
with males being the most affected one. This collection of malignant more particularly brain gliomas into grades I-IV by integrating histo­
tumors of central nervous system actually protrudes from the glial cells pathological and molecular genetic markers (Isocitrate dehydrogense
(which basically surround neurons for support and insulation within the Wildtype /mutation & 1p/19q codeleted /non-codeleted). Due to inva­
brain). Briefly, as per histology, glioma can be classified into high-grade siveness as well as aggressiveness; this undifferentiated Glioblastoma
glioma like anaplastic astrocytoma IDH mutant/IDH wild type, mutant/ multiforme has been designated as Grade IV tumor by WHO [2]. The
IDH wild type, glioblastoma IDH and midline diffuses Glioma H3 current therapeutic regimen for GBM includes safe removal of the lesion
K272M-mutant or low-grade gliomas like astrocytoma, oligoden­ along with chemotherapy with temozolomide (TMZ) and radiotherapy,
droglioma mixed glioma (mixurre of astrocyte and oligodendrocytes). but still limited clinical success had been met [3–5].
Other tumors of glial origin are ependymomas, CNS lymphomas, Epigenetics regulate both formation and suppression of tumors by

* Corresponding authors.
** Corresponding author at: School of Pharmaceutical Sciences, Apeejay Stya University, Gurugram, Haryana, India.
E-mail addresses: fhpottoo@iau.edu.sa, fahihyder@gmail.com (F.H. Pottoo), rxnoushad@gmail.com (Md.N. Javed), fakhan@iau.edu.sa (F.A. Khan).

https://doi.org/10.1016/j.semcancer.2020.04.001
Received 14 November 2019; Received in revised form 7 April 2020; Accepted 8 April 2020
Available online 14 April 2020
1044-579X/© 2020 Elsevier Ltd. All rights reserved.

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

regulating different types of genetic modifications such as, cytosine 3. Interaction of miRNAs with signaling pathways in
methylation at CpG doublets, acetylation, methylation, & phosphory­ glioblastoma multiforme
lation of histone proteins or by change in the three dimensional chro­
matin conformation [3,6]. miRNAs which are non-coding RNAs of 22bp Mutation or amplification of Genes had been commonly reported
size mediate expression of several genes both at mRNA and protein level among p53, receptor tyrosine kinase (RTK)/RAS/PI3K as well as in
[7–9]. miRNA signatures are intricately linked with onset and progres­ retinoblastoma tumor suppressor signaling pathways [25]. The dysre­
sion of GBM, the upregulated miRNAs are miR-10a, miR-22, miR-34a, gulated receptor tyrosine kinase (RTK, including, MET, EGFR, PDGFRα,
miR-129-3p, miR-132, miR-146b-5p, miR-149, miR-152, miR-155, etc.) signaling pathways are correlated for their critical role in pro­
miR-195, miR-221, miR-222, miR-296-3p, miR-671-5p, while gression of various cancers including gliomas, but have now become
down-regulated are let-7b, miR-19a, miR-19b, miR-20a, miR-767-5p, prominent characteristics of GBM cases [26–28]. A number of primary
miR-106a, miR-181a, -301b, miR-505 [10,11]. The miRNAs have GBM based studies have established overexpressed EGFR (epidermal
gained immense interest since their evidence of linkage between altered growth factor receptor) presence in more than 60% of cases [29,30]. The
expression levels and tumors had been reported [12,13]. In GBM, major EGFR signaling pathways from the plasma membrane includes: PI3K/­
molecular targets of miRNAs includes PTEN, MDM2 (Mouse double protein kinase B (PKB/AKT) pathway, protein kinase C (PKC) pathway,
minute 2 homolog), TSC1 (tuberous sclerosis complex-1), POLD2 (DNA RAS/mitogen activated protein kinase (MAPK)/extracellular signal­
Polymerase Delta 2,), TGFβ-RII (transforming growth factor-β receptor –regulated kinase (ERK) pathway and Janus Kinase (JAK)/STAT
II), CTGF (connective tissue growth factor) and CAMTA1 (Calmodulin pathway [31]. miRNAs in accordance with their capacity to modulate
Binding Transcription Activator 1) [14]. The GBM correlates with signaling pathways ensure homeostasis, metastasis and fibrosis [32], e.g
elevated expression levels of ALDH1A3 (Aldehyde Dehydrogenase 1 miR-29a downregulates PTEN, EphB3 and SOX4 to mediate activation of
Family Member A3). Hence, adaptation of approaches, such as miRNA complicated post-transcriptional program for promoting the prolifera­
targeting drugs or biological macromolecules, would provide some tion and invasion of GBM [33]. miR-34a & miR-451 inhibit signaling
unique solutions to inhibit elevated expression of ALDH1 levels in GBM through AKT and thereby exhibit tumor suppressor effects in GBM [34,
and unravel development of potential therapeuticals [15,16]. It is also 35]. miR-7 inhibits EGFR and Akt activity thereby reduces GBM inva­
quite important to confess the fact that even a single miRNA may siveness [36]. Thus, miRNAs exhibit potential role in the clinical man­
regulate multiple targets which would influence the formation, pro­ agement of GBM w.r.t modulation of diverse signaling pathways
gression, proliferation and migration of GBM [17,18]. The expression (Fig. 1).
levels of some IDH mutations (prognostic), promoter methylation,
MGMT (O-methylguanine–DNA methyltransferase) and 1p/19q predic­ 3.1. RAS/mitogen activated protein kinase (MAPK)/extracellular
tive co-deletion, related biomarkers are extensively employed in the signal–regulated kinase (ERK) pathway
clinical management of GBM [19].
EGFR recruits the SH2 domain bearing protein-GRB2. The GRB2 is
2. miRNA Biogenesis associated with RAS GEF SOS as a preformed complex, to facilitate
activation of RAS. This activated RAS drives the RAF-MAPK/ERK kinase
Briefly, biogenesis of miRNA starts with the transcription of primary (MEK)-ERK1/2 signaling cascade. Subsequently, activation of ERK1/2
miRNA (> 1kbps) from the cellular DNA sequence, via RNA polymerase (critical regulator of proliferation, survival and metabolism) phosphor­
II enzyme, in the nucleus of animal cells. Initially it resembles as a long ylates downstream substrates, which get translocated into the nucleus to
hairpin like double stranded RNA structures, comprising multiple modulate various proteins and transcription factors. In tumors with
nucleotide segments required for its processing and maturation. In the mutated or deleted NF1, the activation of RAS was reported which was
nuclear region, an endonuclease i.e. RNAse III Drosha, in association measured in terms of p-ERK and p-MEK [31,37,38].
with cofactor DiGeorge Syndrome Critical Region 8 (DGCR8); develops
into unique complex called microprocessor. This complex precisely cuts 3.2. PI3K/protein kinase B (PKB/AKT) pathway
the pri-miRNA and degradation it into a secondary product of ~65bps
pre-miRNA [20]. Such newly generated pre-miRNAs are then exported Activation of EGFR induces phosphoinositide 3-kinase (PI3K) to
into the cytoplasm via transportation complex comprising exportin 5 synthesize phosphatidylinositol (3,4,5) trisphosphates (PIP3) from
(EXP5), RAN, GTP and pre-miRNA [21,22]. As soon as the complex Phosphatidylinositol 4,5-bisphosphate (PIP2). At the membrane site,
crosses the nuclear membrane, the RNAse protein (Dicer) cleaves the Akt bound to PIP3, is phosphorylated at Thr308PDK1 by PDK1, leading
pre-miRNA into a ~22 bp miRNA creating a small Duplex miRNA with to its partial activation. Subsequently, PDK2 phosphorylates Akt at
the help of TAR RNA-binding protein (TRBP or PACT). Activated Dicer Ser473 to induce full activity [39]. Such duo-phosphorylated Akt in­
as well as DGCR8 complex are believed to be auto-regulated in the nu­ duces the cytoplasmic events like phosphorylation of TSC2. Activated
cleus of the cells. The newly formed Duplex miRNA is laden on a TCS2 relieves repressive effects on Rheb causing cell proliferation, in­
particular AGO protein (1 of a 4 types), forming a precursor-RNA hibition of apoptosis and downstream activation of mTOR [40]. In
Induced silencing complex (pre-RISC). Subsequently, after degradation contrary, tumor suppressor phosphatase and tensin homolog (PTEN)
of one strand of the double helix in pre-RISC it immediately modifies to dephosphorylates PIP3 and inhibits enzymatic activities of Akt (mutated
become mature RISC [18,20,23]. A number of studies have shown in GBM). Recently, it was also established that before the amplification
regulation of hundreds of targets via a single miRNA including glioma of EGFR, there is loss of PTEN containing 10q chromosome [31]. GBM
formation and its proliferation. About 1% of miRNAs (Mirtrons) are tumor and cell lines based genomic analysis revealed mutation in
produced via non-canonical mechanism in which need of Drosha/Dgcr8, RTK/PTEN/PI3K pathway by confirming elevated phospho-AKT levels
in the nucleus of a cell, is avoided. Additionally these types of miRNA are [41,42].
actually encoded by the excited introns of the primary mRNA transcript
which are cleaved by action of spliceosomes as well as endonucleases as 3.3. The JAK/STAT pathway
intron lariat. The intron lariat is debranched or linearized by DBR1
enzyme (in Humans) followed by transportation into cytoplasm by ac­ Upon activation of EGFR, intracellular JAKs phosphorylate each
tion of XPO5 enzyme for DICER cleavage and subsequent maturation other, which inturn phosphorylates STATs, leading to STAT dimeriza­
into miRNA. The miRNA interacts with mRNA either at its 3’ UTR region tion and their translocation into the nucleus [43], followed by their
to suppress or at 5’UTR region to favor expression levels [20,24]. binding at enhancer sequences located in 3’ or 5’ position of dimer or
complex oligomer to halt transcriptional activities of target gene [31,44,

392

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

Fig. 1. Modulation of EGFR signaling pathways in gliobalstoma multiforme with miRNAs.

45].In GBM, although STAT-3 exhibit relatively lower frequency of which favored selective targeting and accumulation of paclitaxel (PTX)
mutation however localized (microenvironment) presence of IL-6 leads liposomes in glioma cells (up-to 4.72-fold higher uptake than pure drug)
to its aberrant activation [46]. [51].
In parallel to this, SLC7A5-13, and SLC7A15 [L-type amino acid
3.4. The Phospholipase C (PLC)/PKC pathway transporters (LATs)] and SLC7A1-4 and SLC7A14 [cationic amino acid
transporters (CATs)] which belong to solute carrier family 7 (SLC7) as
Activated EGFR recruits and activates PLC, which catalytically well as heteromeric amino acid transporter (HAT) such as LAT1
cleavages PIP2 into inositol 1,4,5-trisphosphate (IP3) and di-acyl glyc­ (SLC7A5); exhibit unusual high expression levels during cancer pro­
erol (DAG). Active PLC also stimulates PKC which inturn triggers gression, and were thus visualized as channels delivering essential
numerous regulatory molecules to affect angiogenesis, infiltration, amino acids into the tumor cells via some unknown cancer-associated
proliferation and survival events in tumors [31]. The downstream ef­ reprogrammed metabolic networks, thereby promoting normal cancer
fectors of PKC isoenzymes are cellular cycle regulators- p53 and p21, growth into high-grade tumors with metastasis [52,53]. The, overex­
regulators of cellular growth & proliferation -RAS-RAF1 and glycogen pressed multi-drug transporters like P-glycoprotein at brain capillary
synthase kinase 3 (GSK3), cell motility regulators-integrins, cell survival endothelial cells and astrocytic end-feet, further complicate the situation
regulators- BCL2 and BAD, as well as inflammation regulator NFκB [47, by ensuring expulsion of exogenous molecules/drugs out of brain i.e
48]. reduction of drug bioavailability. Hence, approaches for combining
chemo-therapeuticals with inhibitory substrates of such efflux confer­
4. Rationalizing nanoparticles for miRNA delivery in ring transporters are critical to reduce dose of drugs, increase the
glioblastoma multifome bioavailability as well as to encounter multidrug resistance incidents of
GBM [54].
4.1. The blood-brain barrier and challenges to drug delivery Apart from the protective role as well as secretion of both barrier-
promoting as well as barrier-disrupting factors, BBB, by virtue of its
Tangled transmembrane complex of non-fenestrated endothelial paracrine interaction with astrocytes, pericytes and ECs, enables inter­
cells, microglia, pericytes, astrocytes and basement membrane assem­ linked network of neuronal signals. The paracrine interaction of BBB
bled with structural proteins such as collagen, laminin, junctional with astrocytes co-regulates the blood flow. While astrocytes also
adhesion molecule-1, occludin, claudins, and cytoplasmic proteins regulate matrix metalloproteinase for the purpose of breakdown of the
(zonula occludens-1 & 2) form extensive tight junctions of protective basement membrane during neuronal inflammation, so that entry of
blood-brain barrier (BBB) surrounding the brain. Primary role of BBB is immune cells would be felicitated inside the brain [55]. Similarly,
to strictly deny paracellular entry, majorly of systemically circulating functions of pericytes which enwrap blood microvessels cells, is to
molecules, more prominently hydrophilic agents, owing to their exog­ maintain BBB homeostasis, regeneration of stroma, process of angio­
enous origin [49]. The process of piocytosis which is some non-specific, genesis and neo-vascularization, regulation of antigen presenting cells
non-saturable, non-carrier-mediated vesicular transportation however during brain infections, differentiation of neural stem cell attributes, as
may felicitate vesicular uptake of bulk fluids and their solute compo­ well as EC proliferation [56]. In addition, lecticans, tenascins, hyal­
nents within the cells, from the site of their microenvironment. The uronic acid, proteoglicans as well as hyaluronan constitutes extracel­
passage of these hydrophilic solutes is usually energy-independent lular matrix (ECM) which serves as base component of BBB structures,
processes so their level in brains are insignificant unless transported as and also interacts with other matrix proteins such as laminin and
substrates of any transporters (such as GLUT 1,3,4,5,6 and 8: glucose endothelial integrin, to ensure establishment of harbored endothelium
transporters for hydrophilic glucose molecules) [50]. In order to exploit networks as well as regulation of paracellular diffusion [56,57]
this; the glucose-RGD (Glu-RGD) derivative was synthesized as ligand

393

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

4.2. Impact of glioblastoma multifome progression on the blood-brain nanoparticles (LPNs), efficiently targeted glioblastomas [78].
barrier PLGA-Nanoparticle encapsulated antisense miR-21 delivered prior to
TMZ treatment of cells, significantly brought down the number of viable
In the progression of cancerous states, including GBM, the changes in cells (p < 0.001). Similarly cell cycle arrest at G2/M phase increased
morphology, permeability and functionality of BBB, render it more (1.6-fold) upon treatment with TMZ in U87 MG cells [79]. The delivery
suitable terminology i.e blood-brain tumor barrier (BBTB) [58]. During of anti-miR-21 oligo-nucleotides via Chlorotoxin (CTX)-coupled SNALP
initial (1st phase) progression of cancer, capillaries maintain continuous promoted silencing of miR-21, elevation of PTEN and PDCD4, activation
and non-fenestrated i.e. integrity, morphology and functionality of BBB of caspase 3/7, culminating in reduced tumor size/ proliferation,
is not affected significantly. But, as the cancer progress into 2nd growth apoptosis and improvement of animal survival [80]. Nadiya M Teplyuk
phase, cancer cells start invading surrounding tissues, grow-up into et al., 2016 applied convection-enhanced delivery (CED) to continu­
around few (2-3) mm of volume, followed by alteration in permeability ously deliver anti-miR-10b-containing nanoparticles for two weeks, via
of the BBB due to neovascularization. In this angiogenesis mediated intracranial osmotic pumps, significant reduction in intracranial GBM
neovasculature formation, newly formed capillaries (12 nm size) which progression and cell proliferation with increase in apoptosis was
are of continuous and fenestrated morphology, start allowing very se­ observed. All of these properties were evident from decreased staining
lective and spherical nutrient molecules up-to 12 nm to cross through for proliferation markers PCNA and KI67 (cleaved caspase 3 staining)
this newly formed barrier i.e., BBTB. While, in 3rd or final stage; char­ [81].
acteristic features such as enlargement of capillary fenestration size Polymeric nanogels (NGs)-miR-34a nano-polyplexes upregulated
usually upto 48 nm, reduction in micro-vessel basement membrane miR-34a while downregulating its target oncogenes like c-MET, CDK6,
thickness, loss of junctional proteins, increased inter-endothelial gaps Notch1 and Bcl-2 in Human U-87 MG GBM cells resulting in inhibition of
up-to 1 μm, taunts the full integrity of BBTB [59]. The peptides based proliferation/migration of cells in vitro, and also tumor suppression in
biomolecules are being released from tumor cells of CNS into blood GBM-bearing SCID mice [82]. Dendritic polyglycerolamine (dPG-NH2)–
serum which can be exploited as affluent source of potentially predictive miR-34a polyplex reduced expression of oncogenes like C-MET, CDK6,
cancer specific biomarkers. CTCs, ctDNA and MVs have been sampled Notch1 and BCL-2 in U-87 MG human cells leading to inhibition of cell
from different biofluids in GBM patients [60,61]. cycle progression, proliferation and migration of GBM cells in vitro, and
reduction of tumor growth in GBM inoculated SCID mice [83]. CXCR4
4.3. Nanotechnology enabled miRNA targeted delivery in glioblastoma receptor-stimulated lipoprotein-like nanoparticle (SLNP) loaded with
multifome microRNA-34a in the core, reduced expression of Y-box 2 (sex-de­
termining region) and Notch1, thereby Inhibited GICs stemness and
The complexity of CNS and presence of physiological barriers pos­ chemoresistance and significantly prolonged the survival of GICs-bear­
sesses major scientific challenges, in conventional delivery approaches ing mice [84]. An evident increment in apoptosis and decrement in
for newly synthesized drugs or hydrophilic molecules or phytocon­ proliferation was observed when GBM tissue slices were treated with
stituents in neurological disorders and cancers, such as glioma (GBM) nanoparticle loaded with miR-29b, resulting in reduction in expression
[62–65]. Furthermore, In contrast to the neuronal delivery of synthetic of COL1A2, COL3A1, COL4A1, ELN, ITGA11, MMP24, and SPARC, to
chemical analogues, miRNA-based therapeuticals, bear additional mediate an anticancer effect [85].
challenges due to their susceptibility towards enzyme and PH mediated In gliomas, miR221 is liked with invasiveness and temozolomide
degradation even before reaching the target sites [66]. Hence in lieu of resistance. The, delivery of temozolomide (TMZ) in association of anti-
above, it would be judicious to develop an efficient and biocompatible miR221 PNA, loaded on mesoporous silica nanoparticles (MSNPs),
carrier system which would felicitate entry of nucleic acid loads to induced apoptosis in TMZ resistant T98 G cells [75]. The combination of
specific areas within the brain, without being subjected to degradation. anti-miR-21 oligo-nucleotides via Chlorotoxin (CTX)-coupled SNALP
The nanotechnology fabricated carriers e.g polymeric systems, lipid with Sunitinib (a tyrosine kinase inhibitor) decreased tumor cell pro­
based carriers, liposomes and metallic nanoparticles hold the capacity liferation & enhanced apoptosis in C57BL/6 mice [80]. The delivery of
and efficiency to deliver the miRNA based loads [67–69]. Inaddition, the 5-FU and antisense miR-21 constituting synergistic pharmaceutical
surface of metallic nanoparticles, such as spherical silver and gold NPs, compositions via PAMAM based carriers exhibited prominent anti­
have been attributed to their catalytic properties; hence exploited for proliferative effect in human glioma U251 cells [86]. In association with
functionalization purposes of peptide analogues [70].The integration of TMZ, both antagomiR-21 and antagomiR-10b, delivered into U87MG
nanotechnology to deliver rationalized pharmaceutical compositions, is and Ln229 GBM cells via PLGA based and cRGD-targeting nano-carriers,
the most favorable approach which significantly reduces the burden of not only efficiently arrested cell cycle at G2/M phase but also sensitized
drug regimen by reducing the course of treatment, coupled with cells, even to low concentrations of TMZ [87]. The, combo-delivery of
improved potential therapeutic outcomes and minimal side effects miR-148a and miR-296-5p containing pharmaceutical compositions not
[71–73]. The efficient delivery of tumor suppressor micro­ only inhibited stem cell phenotype of human GBM cells in vitro but also
RNA–conjugated systems using nanotechnological processed carriers in increased long-term survival in athymic nude NCR Nu/Nu and response
brain is very promising field. The miRNA nanoparticles based drug de­ to γ radiation therapy [88]. Among metallic nanoparticles, Iron oxide
livery systems therefore provides new means to rationalize selection of owing to its magnetic properties is being exploited for external magnet
appropriate approaches to enhance overall permeability of loaded bio­ controlled localized release of drugs as well as hyperthermia mediated
molecules or drugs, with enhanced retention impacts i.e. by exploiting apoptosis of tumors, including GBM [89,90]. While, silver and gold
nanoparticulated drug delivery leading into enhanced permeation and nanoparticles, owing to their anti-infectious properties as well as unique
retention (EPR) of loaded miRNA preferentially within the cancer SPR (Surface Plasmon Resonance) mediated analytical signal amplifi­
affected regions in brain for the effective clinical management of GBM cations, are exploited not only for their restricted use as drug carriers but
[74–76]. Therefore, various novel drug delivery approaches for miRNA also for diagnosis of state of GBM [91–93]. Hence, unique single hybrid
delivery in cases of GBM had been developed in the recent past. system of both gold and iron oxide nanoparticles (GIONs) was developed
In order to efficiently knock down miR-21 (oncomir) expression in with functionalized β-cyclodextrin-chitosan (CD-CS) hybrid polymer
GBM, anti-miR-21 LNA loaded Multi-valentfolate (FA)-conjugated 3 W J and PEG-T7 peptide, for co-delivery of miR-100 and antimiR-21, the
RNP were injected into glioblastoma cells in vitro to induce rescue of delivery of which in association of TMZ lead into synergistically
tumor suppressors; PTEN and PDCD4 culminating into apoptosis [77]. enhanced anticancer potentials in U87-MG GBM cell-derived orthotopic
The delivery of miR-21 antisense oligonucleotide (anti-miR-21) via lipid xenograft models in mice [94]. Hence with inspiration of all related
and polymer constituting unique single hybrid systems; lipid-polymer studies; nanotechnology platform enabled delivery of RNA interference

394

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

therapeuticals assures promising therapeutic response to efficiently 6. Conclusion


combat gliomas (Tables 1 and 2).
Herein, we reviewed the biological nature of GBM tumors, the
5. Future prospects devastating outcome for patients, the failure of chemo and radio therapy
and recent progress in establishing the role of miRNAs as diagnostic
The recent promising discoveries and evidences on critical role of markers as well as therapeutic agents (alone or in combination with
miRNAs in the formation & progression of glioma, enable some of them chemotherapeutic molecules) in mitigation of GBM. The deeper under­
to emerge as exclusive clinical hallmarks in diagnosis, prognosis as well standing of miRNAs and their capacity to modulate signaling pathways,
as novel agents in mitigation of glioblastoma multiforme (GBM). Sub­ which govern tumorigenicity opens new avenues for improving the life
sequently, with the progression of GBM; the integrity of BBB is expectancy and QoL of GBM patients. But these miRNAs exhibit low
compromised in a way that these miRNAs appeared to be released in a stability, solubility and achieve non-site selective targeting, which
systemic circulation, which could be exploited for early diagnosis & paved way for nanotechnological based novel delivery approaches. Over
identification of different stages of giloma. Advanced analytical tech­ the last few years, progress and advancement in nanotechnology-driven
niques like northern blotting, qRT-PCR, microarrays and NGS needs to strategies to fabricate novel material/s for targeted drug delivery of
be standardized, validated and integrated to generate and process the biological macromolecules (such as peptides, RNA, miRNAs etc) and
complex raw data in establishing definitive clinical decisions. Further­ chemotherapeuticals enabled by the state of the art, controlled, func­
more, additional techniques such as enzymatic amplification may be tionalized and target centric modus operandi, accomplishes specific
required to identify those miRNAs which are specific hallmarks in GBM functions to cater the need required for. Different types of nanoparticles,
but give poor signaling. The unique SPR attributes of metallic nano­ such as silver nanoparticles, colloidal gold, silica nanoparticles, lipo­
particles would favor adequate amplification of these detection signals. some, polymeric carriers, bioinspired systems, niosomes, limicubes,
In the past assessment of promising role of nanotechnology based ma­ NLC, SLN etc, have been rationalized for optimal benefit in different
terials and processes with synthetic and spiked- samples validated as pathological states including neuronal cancers. These delivery systems
proof-of-principle. But the absence of internationally recognized regu­ not only protect miRNAs from degradation by nucleases but also in­
latory procedures as well as stringent quality control strategies limits the creases their half-life in the blood or serum. Inaddition, numerous
clinical translation of these strategies. The above mentioned unmet is­ clinical trials reported combination therapies, likely be the most prom­
sues and challenges needs to be addressed in a very judicious and sci­ ising method for GBM treatment, and that nanotechnology allows
entific way which would regulate the integration of nanotechnological conjugation of miRNAs with anti-cancer drugs and that too with syn­
techniques for site specific delivery of miRNAs in clinical settings. ergistic effects. Thus the successful treatment of GBM lies with the
effective nano-tech enabled delivery of miRNAs across the BBTB either
alone or in combinations.

Table 1
Nanotechnology enabled miRNA targeted delivery in Glioblastoma Multiforme (In vitro studies).
S. Nanoformulation/s type Rationale Model (animal/cell Outcomes References
NO line)

1. anti-miR-21 LNA loaded Multi- miR-21 knock down U87EGFRvIII or Gli36 Glioblastoma cell apoptosis [77]
valentfolate (FA)-conjugated 3 W J cells
RNP
2. Chlorotoxin (CTX)-coupled SNALP- miR-21 silencing, U87 human GBM and Decrease in tumor cell proliferation [95]
anti-miR-21 oligonucleotides PTEN and PDCD4 upregulation, caspase GL261 mouse glioma
3/7 activation cells
3. PACE-antimiR-21 nanoparticles & miR-21 suppression, PTEN upregulation U87 cells Apoptosis of human GBM cells [96]
PLA-HPG- antimiR-21
nanoparticles
4. PLGA nanoparticle encapsulated knock down endogenous miR-21 U87 MG, LN229, and Overexpression of the miR-21 target genes [79]
antisense miR-21 T98 G cells PTEN (by 67%) and caspase-3 (by 15%) upon
cotreatment with TMZ
5. 5-FU-Loaded PAMAM- antisense- Down-regulation of miR-21 Human glioma U251 as-miR-21 significantly improved the [86]
miR-21 cells cytotoxicity of 5-FU and dramatically increased
the apoptosis of U251 cells
6. LPNs loaded with pemetrexed and Co-delivery of anti-miR-21 & U87MG human improved accumulation of LPNs in the nucleus [78]
anti-miR-21 pemetrexed glioblastoma cells. of U87MG cells.
7. Nanoparticle loaded miR-29b marked decrease in proliferation and GBM tissue slices miR-29b inhibits the expression of COL1A2, [85]
significant increase in apoptosis after COL3A1, COL4A1, ELN, ITGA11, MMP24, and
miR-29b treatment SPARC, which mediate an anticancer effect.
8. NG-miR-34a nano-polyplexes upregulation of miR-34a and Human U-87 MG GBM Inhibition of cell proliferation and migration [82]
downregulation of its target oncogenes cells
like c-MET, CDK6, Notch1 and Bcl-2.
9. dPG-NH2–miR-34a polyplex lower expression of C-MET, CDK6, U-87 MG human cells Inhibition of cell cycle progression/ [83]
Notch1 and BCL-2 genes proliferation and migration of GBM cells
10. Cy5-MSNPs loaded with anti- down-regulation of miR221 temozolomide-resistant Effective induction of apoptosis (70.9% of [75]
miR221 PNA T98 G cell line. apoptotic cells)
11. cRGD-targeted PLGA nanoparticles Co-inhibition of miR-21 and miR-10b U87MG and Ln229 Improvement in sensitivity of these cells to [87]
encapsulating antagomiR-21 and increased cell cycle arrest at G2/M GBM cells lower concentrations of TMZ
antagomiR-10b phase upon TMZ treatment.
12. nano-miRs containing miR- Delivery of miR-148a + miR-296-5p GBM1A cells Inhibit the stem cell phenotype of human GBM [88]
148a + miR-296-5p cells in vitro

Abbreviations: Mesoporous silica nanoparticles (MSNPs; temozolomide (TMZ; peptide nucleic acids (PNAs; poly(amine-co-esters(PACE); poly(lactic acidand hyper­
branched polyglycerol (PLA-HPG); lipid-polymer hybrid nanoparticles (LPNs; Chlorotoxin (CTX; polymeric nanogels (NGs; Poly(lactic-co-glycolic acid(PLGA); Locked
Nucleic Acids (LNAs

395

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

Table 2
Nanotechnology enabled miRNA targeted delivery in Glioblastoma Multiforme (In vivo studies).
S. Nanoformulation/s type Rationale Model (animal/cell line) Outcomes References
NO

1 ApoE coated PACE-antimiR -21 NPs & miR-21 inhibition, Male RNU rats (Convection-enhanced Increase in PTEN expression [96]
PLA-HPG- antimiR-21 nanoparticles delivery)
2. anti-miR-21 LNA loaded Multi-valentfolate PTEN and PDCD4 Athymic nu/nu outbred mice Tumor growth regression [77]
(FA)-conjugated 3 W J RNP upregulation
3. Chlorotoxin (CTX)-coupled SNALP-anti- miR-21 silencing Adult male C57BL/6 mice Decreased tumor cell proliferation & [80]
miR-21 oligonucleotides enhanced apoptosis in combination
with tyrosine kinase inhibitor;Sunitinib
4. Co-delivery of miR-148a and miR-296-5p Delivery of miR- athymic nude NCR Nu/Nu mice Long-term survival [88]
148a + miR-296-5p
5. Nanogels (NGs) -miR-34a nano-polyplexes GBM-bearing SCID mice Inhibited tumor growth [82]
6. cRGD-targeted and non-targeted PLGA Co-inhibition of miR-21 nude mice (nu/nu) Increase in cellular chemosensitivity [87]
nanoparticles encapsulating antagomiR-21 and miR-10b towards lower doses of TMZ
and antagomiR-10b
7. dPG-NH2–miR-34a polyplex Upregulaion of miR-34a SCID mice Inhibition of tumor growth [83]
8. FH38-miR34a-SLNPs Reduces sex-determining GICs-derived orthotopic mice models Inhibits GICs stemness and [84]
region Y-box 2 and chemoresistanceand significantly
Notch1 expression prolongs the survival of GICs-bearing
mice.
9. anti-miR-10b-containing nanoparticles Downregulation of miR- Intracranial human GSC-derived Attenuated growth and progression of [81]
10b (oncogenic miRNA) xenograft and murine GL261 allograft established intracranial GBM
models in athymic and
immunocompetent mice.
10. CD-CS coated GIONs co-loaded with miR- Downregulate miR-21 U87-MG GBM cell-derived orthotopic Significant increase in survival of mice [94]
100 and antimiR-21. surface and upregulate miR-100 xenograft models in mice. co-treated with T7-polyGIONs loaded
functionalization with PEG-T7 peptide with miR-100/antimiR-21 plus
using CD-adamantane host-guest systemic TMZ
chemistry.

Abbreviations: Chlorotoxin (CTX)-coupled (targeted) stable nucleic acid lipid particle (SNALP); glioma-initiating stem-like cells (GSC); gold-iron oxide nanoparticles
(polyGIONs); β-cyclodextrin-chitosan (CD-CS); cationic poly(amine-co-ester) (PACE); poly(lactic acid) and hyperbranched polyglycerol (PLA-HPG).; peptide nucleic
acid (PNA); convection-enhanced delivery (CED)

Declaration of Competing Interest Glioblastoma: The Role of Extracellular Transfer, Stem Cell Reports. 8 (2017)
1497–1505, https://doi.org/10.1016/j.stemcr.2017.04.024.
[12] T.K. Hinz, L.E. Heasley, Translating mesothelioma molecular genomics and
The authors declare no conflict of interest dependencies into precision oncology-based therapies, Semin. Cancer Biol. (2019),
https://doi.org/10.1016/j.semcancer.2019.09.014.
References [13] R.C. Lee, R.L. Feinbaum, V. Ambros, The C. elegans heterochronic gene lin-4
encodes small RNAs with antisense complementarity to lin-14, Cell 75 (1993)
843–854, https://doi.org/10.1016/0092-8674(93)90529-Y.
[1] M. Weller, W. Wick, K. Aldape, M. Brada, M. Berger, S.M. Pfister, R. Nishikawa, [14] E.A. Toraih, N.M. Aly, H.Y. Abdallah, S.A. Al-Qahtani, A.A. Shaalan, M.H. Hussein,
M. Rosenthal, P.Y. Wen, R. Stupp, G. Reifenberger Glioma, Nat Rev Dis Primers. 1 M.S. Fawzy, MicroRNA-target cross-talks: Key players in glioblastoma multiforme,
(2015) 15017, https://doi.org/10.1038/nrdp.2015.17. Tumour Biol. 39 (2017), https://doi.org/10.1177/1010428317726842,
[2] F. Hanif, K. Muzaffar, K. Perveen, S.M. Malhi, S.U. Simjee, Glioblastoma 1010428317726842.
Multiforme: A Review of its Epidemiology and Pathogenesis through Clinical [15] G. Li, Y. Li, X. Liu, Z. Wang, C. Zhang, F. Wu, H. Jiang, W. Zhang, Z. Bao, Y. Wang,
Presentation and Treatment, Asian Pac J Cancer Prev 18 (2017) 3–9, https://doi. J. Cai, L. Zhao, U.D. Kahlert, T. Jiang, W. Zhang, ALDH1A3 induces mesenchymal
org/10.22034/APJCP.2017.18.1.3. differentiation and serves as a predictor for survival in glioblastoma, Cell Death
[3] B. Banelli, A. Forlani, G. Allemanni, A. Morabito, M.P. Pistillo, M. Romani, Dis. 9 (2018), https://doi.org/10.1038/s41419-018-1232-3.
MicroRNA in Glioblastoma: An Overview, International Journal of Genomics. [16] W. Zhang, Y. Liu, H. Hu, H. Huang, Z. Bao, P. Yang, Y. Wang, G. You, W. Yan,
(2017), https://doi.org/10.1155/2017/7639084. T. Jiang, J. Wang, W. Zhang, ALDH1A3: A Marker of Mesenchymal Phenotype in
[4] Z. Cheng, H.Z. Wang, X. Li, Z. Wu, Y. Han, Y. Li, G. Chen, X. Xie, Y. Huang, Z. Du, Gliomas Associated with Cell Invasion, PLOS ONE. 10 (2015) e0142856, https://
Y. Zhou, MicroRNA-184 inhibits cell proliferation and invasion, and specifically doi.org/10.1371/journal.pone.0142856.
targets TNFAIP2 in Glioma, Journal of Experimental & Clinical Cancer Research. [17] A.M. Abdelfattah, C. Park, M.Y. Choi, Update on non-canonical microRNAs,
34 (2015) 27, https://doi.org/10.1186/s13046-015-0142-9. Biomolecular Concepts. 5 (2014) 275–287, https://doi.org/10.1515/bmc-2014-
[5] G.E.D. Petrescu, A.A. Sabo, L.I. Torsin, G.A. Calin, M.P. Dragomir, MicroRNA based 0012.
theranostics for brain cancer: basic principles, J Exp Clin Cancer Res. 38 (2019), [18] G. Rorbach, O. Unold, B.M. Konopka, Distinguishing mirtrons from canonical
https://doi.org/10.1186/s13046-019-1180-5. miRNAs with data exploration and machine learning methods, Sci Rep. 8 (2018)
[6] M.L. Goodenberger, R.B. Jenkins, Genetics of adult glioma, Cancer Genet. 205 7560, https://doi.org/10.1038/s41598-018-25578-3.
(2012) 613–621, https://doi.org/10.1016/j.cancergen.2012.10.009. [19] R.J. Molenaar, D. Verbaan, S. Lamba, C. Zanon, J.W.M. Jeuken, S.H.E. Boots-
[7] S. Oliveto, M. Mancino, N. Manfrini, S. Biffo, Role of microRNAs in translation Sprenger, P. Wesseling, T.J.M. Hulsebos, D. Troost, A.A. van Tilborg, S. Leenstra,
regulation and cancer, World J Biol Chem. 8 (2017) 45–56, https://doi.org/ W.P. Vandertop, A. Bardelli, C.J.F. van Noorden, F.E. Bleeker, The combination of
10.4331/wjbc.v8.i1.45. IDH1 mutations and MGMT methylation status predicts survival in glioblastoma
[8] J. Sana, P. Busek, P. Fadrus, A. Besse, L. Radova, M. Vecera, S. Reguli, L. better than either IDH1 or MGMT alone, Neuro Oncol. 16 (2014) 1263–1273,
S. Sromova, M. Hilser, R. Lipina, R. Lakomy, L. Kren, M. Smrcka, A. Sedo, O. Slaby, https://doi.org/10.1093/neuonc/nou005.
Identification of microRNAs differentially expressed in glioblastoma stem-like cells [20] M. Ha, V.N. Kim, Regulation of microRNA biogenesis, Nat. Rev. Mol. Cell Biol. 15
and their association with patient survival, Sci Rep. 8 (2018) 1–11, https://doi. (2014) 509–524, https://doi.org/10.1038/nrm3838.
org/10.1038/s41598-018-20929-6. [21] E. Lund, S. Güttinger, A. Calado, J.E. Dahlberg, U. Kutay, Nuclear Export of
[9] S. Shafi, S. Khan, F. Hoda, F. Fayaz, A. Singh, M.A. Khan, R. Ali, F.H. Pottoo, MicroRNA Precursors, Science 303 (2004) 95–98, https://doi.org/10.1126/
S. Tariq, A.K. Najmi, Decoding novel mechanisms and emerging therapeutic science.1090599.
strategies in breast cancer resistance, Curr. Drug Metab. (2020), https://doi.org/ [22] R. Yi, Y. Qin, I.G. Macara, B.R. Cullen, Exportin-5 mediates the nuclear export of
10.2174/1389200221666200303124946. pre-microRNAs and short hairpin RNAs, Genes Dev. 17 (2003) 3011–3016, https://
[10] S. Huang, N. Ali, L. Zhong, J. Shi, MicroRNAs as biomarkers for human doi.org/10.1101/gad.1158803.
glioblastoma: progress and potential, Acta Pharmacol Sin. 39 (2018) 1405–1413, [23] D.-D. Cao, L. Li, W.-Y. Chan, MicroRNAs: Key Regulators in the Central Nervous
https://doi.org/10.1038/aps.2017.173. System and Their Implication in Neurological Diseases, International Journal of
[11] J. Godlewski, R. Ferrer-Luna, A.K. Rooj, M. Mineo, F. Ricklefs, Y.S. Takeda, M. Molecular Sciences. 17 (2016) 842, https://doi.org/10.3390/ijms17060842.
O. Nowicki, E. Salińska, I. Nakano, H. Lee, R. Weissleder, R. Beroukhim, E.
A. Chiocca, A. Bronisz, MicroRNA Signatures and Molecular Subtypes of

396

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

[24] J.P. Broughton, M.T. Lovci, J.L. Huang, G.W. Yeo, A.E. Pasquinelli, Pairing beyond [49] F.H. Pottoo, S. Sharma, M.N. Javed, M.A. Barkat, null Harshita, M.S. Alam, M.
the Seed Supports MicroRNA Targeting Specificity, Mol. Cell. 64 (2016) 320–333, J. Naim, O. Alam, M.A. Ansari, G.E. Barreto, G.M. Ashraf, Lipid-based
https://doi.org/10.1016/j.molcel.2016.09.004. nanoformulations in the treatment of neurological disorders, Drug Metab. Rev. 52
[25] F. Cheng, D. Guo, MET in glioma: signaling pathways and targeted therapies, (2020) 185–204, https://doi.org/10.1080/03602532.2020.1726942.
Journal of Experimental & Clinical Cancer Research. 38 (2019) 270, https://doi. [50] Null Harshita, M.A. Barkat, S.S. Das, F.H. Pottoo, S. Beg, Z. Rahman, LIPID-BASED
org/10.1186/s13046-019-1269-x. NANOSYSTEM AS INTELLIGENT CARRIERS FOR VERSATILE DRUG DELIVERY
[26] F. Nawaz, O. Alam, A. Perwez, M.A. Rizvi, M.J. Naim, N. Siddiqui, F.H. Pottoo, APPLICATIONS, Curr. Pharm. Des. (2020), https://doi.org/10.2174/
M. Jha, 3’-(4-(Benzyloxy)phenyl)-1’-phenyl-5-(heteroaryl/aryl)-3,4-dihydro- 1381612826666200206094529.
1’H,2H-[3,4’-bipyrazole]-2-carboxamides as EGFR kinase inhibitors: Synthesis, [51] Q. Fu, Y. Zhao, Z. Yang, Q. Yue, W. Xiao, Y. Chen, Y. Yang, L. Guo, Y. Wu,
anticancer evaluation, and molecular docking studies, Arch. Pharm. (Weinheim). Liposomes actively recognizing the glucose transporter GLUT1 and integrin αvβ3
(2020) e1900262, https://doi.org/10.1002/ardp.201900262. for dual-targeting of glioma, Archiv Der Pharmazie. 352 (2019) 1800219, https://
[27] E. Carrasco-García, M. Saceda, I. Martínez-Lacaci, Role of Receptor Tyrosine doi.org/10.1002/ardp.201800219.
Kinases and Their Ligands in Glioblastoma, Cells 3 (2014) 199–235, https://doi. [52] X. Lu, The Role of Large Neutral Amino Acid Transporter (LAT1) in Cancer, Curr
org/10.3390/cells3020199. Cancer Drug Targets. 19 (2019) 863–876, https://doi.org/10.2174/
[28] D.G.T. Parambi, K.M. Noorulla, M.D. Sahab Uddin, B. Mathew, Epidermal Growth 1568009619666190802135714.
Factor Receptor: Promising Targets for Non-Small-Cell Lung Cancer, in: [53] M. Scalise, M. Galluccio, L. Console, L. Pochini, C. Indiveri, The Human SLC7A5
S. Chakraborti, N.L. Parinandi, R. Ghosh, N.K. Ganguly, T. Chakraborti (Eds.), (LAT1): The Intriguing Histidine/Large Neutral Amino Acid Transporter and Its
Oxidative Stress in Lung Diseases: Volume 2, Springer, Singapore, 2020, Relevance to Human Health, Front Chem. 6 (2018), https://doi.org/10.3389/
pp. 465–471, https://doi.org/10.1007/978-981-32-9366-3_21. fchem.2018.00243.
[29] J. Novakova, O. Slaby, R. Vyzula, J. Michalek, MicroRNA involvement in [54] W. Löscher, H. Potschka, Role of drug efflux transporters in the brain for drug
glioblastoma pathogenesis, Biochemical and Biophysical Research disposition and treatment of brain diseases, Prog. Neurobiol. 76 (2005) 22–76,
Communications. 386 (2009) 1–5, https://doi.org/10.1016/j.bbrc.2009.06.034. https://doi.org/10.1016/j.pneurobio.2005.04.006.
[30] Y. TIAN, Y. NAN, L. HAN, A. ZHANG, G. WANG, Z. JIA, J. HAO, P. PU, Y. ZHONG, [55] J.M. Herndon, M.E. Tome, T.P. Davis, Chapter 9 - Development and Maintenance
C. KANG, MicroRNA miR-451 downregulates the PI3K/AKT pathway through of the Blood–Brain Barrier, in: L.R. Caplan, J. Biller, M.C. Leary, E.H. Lo, A.
CAB39 in human glioma, Int J Oncol. 40 (2011) 1105–1112, https://doi.org/ J. Thomas, M. Yenari, J.H. Zhang (Eds.), Primer on Cerebrovascular Diseases,
10.3892/ijo.2011.1306. Second Edition, Academic Press, San Diego, 2017, pp. 51–56, https://doi.org/
[31] Z. An, O. Aksoy, T. Zheng, Q.-W. Fan, W.A. Weiss, Epidermal growth factor 10.1016/B978-0-12-803058-5.00009-6.
receptor and EGFRvIII in glioblastoma: signaling pathways and targeted therapies, [56] R. Cabezas, M. Ávila, J. Gonzalez, R.S. El-Bachá, E. Báez, L.M. García-Segura, J.
Oncogene 37 (2018) 1561–1575, https://doi.org/10.1038/s41388-017-0045-7. C. Jurado Coronel, F. Capani, G.P. Cardona-Gomez, G.E. Barreto, Astrocytic
[32] M. Inui, G. Martello, S. Piccolo, MicroRNA control of signal transduction, Nat Rev modulation of blood brain barrier: perspectives on Parkinson’s disease, Front Cell
Mol Cell Biol. 11 (2010) 252–263, https://doi.org/10.1038/nrm2868. Neurosci. 8 (2014), https://doi.org/10.3389/fncel.2014.00211.
[33] Y. Zhao, W. Huang, T.-M. Kim, Y. Jung, L.G. Menon, H. Xing, H. Li, R.S. Carroll, P. [57] S. Gupta, S. Dhanda, R. Sandhir, 2 - Anatomy and physiology of blood-brain
J. Park, H.W. Yang, M.D. Johnson, MicroRNA-29a activates a multi-component barrier, in: H. Gao, X. Gao (Eds.), Brain Targeted Drug Delivery System, Academic
growth and invasion program in glioblastoma, Journal of Experimental & Clinical Press, 2019, pp. 7–31, https://doi.org/10.1016/B978-0-12-814001-7.00002-0.
Cancer Research. 38 (2019) 36, https://doi.org/10.1186/s13046-019-1026-1. [58] S.W. Schneider, T. Ludwig, L. Tatenhorst, S. Braune, H. Oberleithner, V. Senner,
[34] Y. Li, F. Guessous, Y. Zhang, C. DiPierro, B. Kefas, E. Johnson, L. Marcinkiewicz, W. Paulus, Glioblastoma cells release factors that disrupt blood-brain barrier
J. Jiang, Y. Yang, T.D. Schmittgen, B. Lopes, D. Schiff, B. Purow, R. Abounader, features, Acta Neuropathol. 107 (2004) 272–276, https://doi.org/10.1007/
MicroRNA-34a Inhibits Glioblastoma Growth by Targeting Multiple Oncogenes, s00401-003-0810-2.
Cancer Res. 69 (2009) 7569–7576, https://doi.org/10.1158/0008-5472.CAN-09- [59] L.G. Dubois, L. Campanati, C. Righy, I. D’Andrea-Meira, T.C.L. de, S.E. Spohr,
0529. I. Porto-Carreiro, C.M. Pereira, J. Balça-Silva, S.A. Kahn, M.F. DosSantos, M. de, A.
[35] D. Yin, S. Ogawa, N. Kawamata, A. Leiter, M. Ham, D. Li, N.B. Doan, J.W. Said, K. R. Oliveira, A. Ximenes-da-Silva, M.C. Lopes, E. Faveret, E.L. Gasparetto, V. Moura-
L. Black, H.P. Koeffler, miR-34a functions as a tumor suppressor modulating EGFR Neto, Gliomas and the vascular fragility of the blood brain barrier, Front. Cell.
in glioblastoma multiforme, Oncogene 32 (2013) 1155–1163, https://doi.org/ Neurosci. 8 (2014), https://doi.org/10.3389/fncel.2014.00418.
10.1038/onc.2012.132. [60] J. Müller Bark, A. Kulasinghe, B. Chua, B.W. Day, C. Punyadeera, Circulating
[36] B. Kefas, J. Godlewski, L. Comeau, Y. Li, R. Abounader, M. Hawkinson, J. Lee, biomarkers in patients with glioblastoma, Br. J. Cancer. (2019), https://doi.org/
H. Fine, E.A. Chiocca, S. Lawler, B. Purow, microRNA-7 Inhibits the Epidermal 10.1038/s41416-019-0603-6.
Growth Factor Receptor and the Akt Pathway and Is Down-regulated in [61] M.A. Zachariah, J.P. Oliveira-Costa, B.S. Carter, S.L. Stott, B.V. Nahed, Blood-based
Glioblastoma, Cancer Res. 68 (2008) 3566–3572, https://doi.org/10.1158/0008- biomarkers for the diagnosis and monitoring of gliomas, Neuro Oncol. 20 (2018)
5472.CAN-07-6639. 1155–1161, https://doi.org/10.1093/neuonc/noy074.
[37] L. Santarpia, S.M. Lippman, A.K. El-Naggar, Targeting the MAPK–RAS–RAF [62] D.M. Teleanu, C. Chircov, A.M. Grumezescu, R.I. Teleanu, Neuronanomedicine: An
signaling pathway in cancer therapy, Expert Opinion on Therapeutic Targets. 16 Up-to-Date Overview, Pharmaceutics 11 (2019), https://doi.org/10.3390/
(2012) 103–119, https://doi.org/10.1517/14728222.2011.645805. pharmaceutics11030101.
[38] J.M. Shields, K. Pruitt, A. McFall, A. Shaub, C.J. Der, Understanding Ras: ‘it ain’t [63] R.K. Upadhyay, Drug Delivery Systems, CNS Protection, and the Blood Brain
over’ til it’s over’, Trends in Cell Biology. 10 (2000) 147–154, https://doi.org/ Barrier, Biomed Res Int. 2014 (2014), https://doi.org/10.1155/2014/869269.
10.1016/S0962-8924(00)01740-2. [64] M.A. Ansari, I.-M. Chung, G. Rajakumar, M.A. Alzohairy, M.N. Alomary,
[39] B.A. Hemmings, D.F. Restuccia, PI3K-PKB/Akt Pathway, Cold Spring Harb Perspect M. Thiruvengadam, F.H. Pottoo, N. Ahmad, A Current Nanoparticles approaches in
Biol. 4 (2012), https://doi.org/10.1101/cshperspect.a011189. Nose to Brain Drug Delivery and Anticancer Therapy - A Review, Curr. Pharm. Des.
[40] F.H. Pottoo, Md. A. Barkat, M.A. Harshita, Md. Ansari, N. Javed, Q.M. Sajid Jamal, (2020), https://doi.org/10.2174/1381612826666200116153912.
M.A. Kamal, Nanotechnological based miRNA intervention in the therapeutic [65] M.A. Ansari, K.F. Badrealam, A. Alam, S. Tufail, G. Khalique, M.J. Equbal, M.
management of neuroblastoma, Seminars in Cancer Biology. (2019), https://doi. A. Alzohairy, A. Almatroudi, M.N. Alomary, F.H. Pottoo, Recent Nano-based
org/10.1016/j.semcancer.2019.09.017. therapeutic intervention of Bioactive Sesquiterpenes: Prospects in cancer
[41] E. Majewska, M. Szeliga, AKT/GSK3β Signaling in Glioblastoma, Neurochem. Res. therapeutics, Curr. Pharm. Des. (2020), https://doi.org/10.2174/
42 (2017) 918–924, https://doi.org/10.1007/s11064-016-2044-4. 1381612826666200116151522.
[42] K.A. McDowell, G.J.R, G.L. Gallia, Targeting the AKT Pathway in Glioblastoma, [66] M.N. Javed, K. Kohli, S. Amin, Risk Assessment Integrated QbD Approach for
Current Pharmaceutical Design, 2011 (accessed November 12, 2019). http://www. Development of Optimized Bicontinuous Mucoadhesive Limicubes for Oral
eurekaselect.com/75009/article. Delivery of Rosuvastatin, AAPS PharmSciTech. 19 (2018) 1377–1391, https://doi.
[43] J.J. O’Shea, D.M. Schwartz, A.V. Villarino, M. Gadina, I.B. McInnes, A. Laurence, org/10.1208/s12249-018-0951-1.
The JAK-STAT pathway: impact on human disease and therapeutic intervention, [67] S. Mishra, S. Sharma, M.N. Javed, F.H. Pottoo, M. Abul Barkat, Null Harshita,
Annu. Rev. Med. 66 (2015) 311–328, https://doi.org/10.1146/annurev-med- M. Amir, M. Sarfaroz, Bioinspired Nanocomposites: Applications In Disease
051113-024537. Diagnosis And Treatment, Pharm Nanotechnol. (2019), https://doi.org/10.2174/
[44] D.A. Harrison, The JAK/STAT Pathway, Cold Spring Harb Perspect Biol. 4 (2012) 2211738507666190425121509.
a011205, https://doi.org/10.1101/cshperspect.a011205. [68] S. Sharma, M. Noushad Javed, F. Pottoo, S. Arman Rabbani, M.D. Barkat, H. Ab,
[45] J.S. Rawlings, K.M. Rosler, D.A. Harrison, The JAK/STAT signaling pathway, D. Sarafroz, M. Amir, Bioresponse Inspired Nanomaterials for Targeted Drug And
Journal of Cell Science. 117 (2004) 1281–1283, https://doi.org/10.1242/ Gene Delivery, 2019, https://doi.org/10.2174/2211738507666190429103814.
jcs.00963. [69] N. Zahin, R. Anwar, D. Tewari, Md.T. Kabir, A. Sajid, B. Mathew, Md.S. Uddin,
[46] B.C. McFarland, J.-Y. Ma, C.P. Langford, G.Y. Gillespie, H. Yu, Y. Zheng, S. L. Aleya, M.M. Abdel-Daim, Nanoparticles and its biomedical applications in health
E. Nozell, D. Huszar, E.N. Benveniste, Therapeutic potential of AZD1480 for the and diseases: special focus on drug delivery, Environ Sci Pollut Res. (2019),
treatment of human glioblastoma, Mol. Cancer Ther. 10 (2011) 2384–2393, https://doi.org/10.1007/s11356-019-05211-0.
https://doi.org/10.1158/1535-7163.MCT-11-0480. [70] M.S. Alam, A. Garg, F.H. Pottoo, M.K. Saifullah, A.I. Tareq, O. Manzoor,
[47] A. do Carmo, J. Balça-Silva, D. Matias, M. Lopes, PKC signaling in glioblastoma, M. Mohsin, M.N. Javed, Gum ghatti mediated, one pot green synthesis of optimized
Cancer Biology & Therapy. 14 (2013) 287–294, https://doi.org/10.4161/ gold nanoparticles: Investigation of process-variables impact using Box-Behnken
cbt.23615. based statistical design, Int. J. Biol. Macromol. 104 (2017) 758–767, https://doi.
[48] R. Garg, L.G. Benedetti, M.B. Abera, H. Wang, M. Abba, M.G. Kazanietz, Protein org/10.1016/j.ijbiomac.2017.05.129.
kinase C and cancer: what we know and what we do not, Oncogene 33 (2014) [71] N. Ahmad, R. Ahmad, M.A. Alam, F.J. Ahmad, M. Amir, F.H. Pottoo, M. Sarafroz,
5225–5237, https://doi.org/10.1038/onc.2013.524. M. Jafar, K. Umar, Daunorubicin oral bioavailability enhancement by surface
coated natural biodegradable macromolecule chitosan based polymeric

397

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.
F.H. Pottoo et al. Seminars in Cancer Biology 69 (2021) 391–398

nanoparticles, Int. J. Biol. Macromol. 128 (2019) 825–838, https://doi.org/ Like miRNA Delivery Nanostructure Suppresses Glioma Stemness and Drug
10.1016/j.ijbiomac.2019.01.142. Resistance through Receptor-Stimulated Macropinocytosis, Advanced Science
[72] N. Ahmad, R. Ahmad, A. Al-Qudaihi, S.E. Alaseel, I.Z. Fita, M.S. Khalid, F. (2020) 1903290, https://doi.org/10.1002/advs.201903290, n/a.
H. Pottoo, Preparation of a novel curcumin nanoemulsion by ultrasonication and [85] J. Shin, H.G. Shim, T. Hwang, H. Kim, S.-H. Kang, Y.-S. Dho, S.-H. Park, C.-K.S, S.
its comparative effects in wound healing and the treatment of inflammation, RSC J. Kim, C.-K. Park, Restoration of miR-29b exerts anti-cancer effects on
Adv. 9 (2019) 20192–20206, https://doi.org/10.1039/C9RA03102B. glioblastoma, Cancer Cell International. 17 (2017) 104, https://doi.org/10.1186/
[73] N. Ahmad, R. Ahmad, A. Al-Qudaihi, S.E. Alaseel, I.Z. Fita, M.S. Khalid, F. s12935-017-0476-9.
H. Pottoo, S.R. Bolla, A novel self-nanoemulsifying drug delivery system for [86] Y. Ren, C.-S. Kang, X.-B. Yuan, X. Zhou, P. Xu, L. Han, G.X. Wang, Z. Jia, Y. Zhong,
curcumin used in the treatment of wound healing and inflammation, 3 Biotech 9 S. Yu, J. Sheng, P.-Y. Pu, Co-delivery of as-miR-21 and 5-FU by poly(amidoamine)
(2019) 360, https://doi.org/10.1007/s13205-019-1885-3. dendrimer attenuates human glioma cell growth in vitro, J Biomater Sci Polym Ed
[74] A. Ambruosi, A.S. Khalansky, H. Yamamoto, S.E. Gelperina, D.J. Begley, J. Kreuter, 21 (2010) 303–314, https://doi.org/10.1163/156856209X415828.
Biodistribution of polysorbate 80-coated doxorubicin-loaded [14C]-poly(butyl [87] M. Malhotra, T.V. Sekar, J.S. Ananta, R. Devulapally, R. Afjei, H.A. Babikir,
cyanoacrylate) nanoparticles after intravenous administration to glioblastoma- R. Paulmurugan, T.F. Massoud, Targeted nanoparticle delivery of therapeutic
bearing rats, Journal of Drug Targeting. 14 (2006) 97–105, https://doi.org/ antisense microRNAs presensitizes glioblastoma cells to lower effective doses of
10.1080/10611860600636135. temozolomide in vitro and in a mouse model, Oncotarget 9 (2018) 21478–21494,
[75] A. Bertucci, E.A. Prasetyanto, D. Septiadi, A. Manicardi, E. Brognara, R. Gambari, https://doi.org/10.18632/oncotarget.25135.
R. Corradini, L.D. Cola, Combined Delivery of Temozolomide and Anti-miR221 [88] H. Lopez-Bertoni, K.L. Kozielski, Y. Rui, B. Lal, H. Vaughan, D.R. Wilson,
PNA Using Mesoporous Silica Nanoparticles Induces Apoptosis in Resistant Glioma N. Mihelson, C.G. Eberhart, J. Laterra, J.J. Green, Bioreducible Polymeric
Cells, Small 11 (2015) 5687–5695, https://doi.org/10.1002/smll.201500540. Nanoparticles Containing Multiplexed Cancer Stem Cell Regulating miRNAs Inhibit
[76] O. van Tellingen, B. Yetkin-Arik, M.C. de Gooijer, P. Wesseling, T. Wurdinger, H. Glioblastoma Growth and Prolong Survival, Nano Lett. 18 (2018) 4086–4094,
E. de Vries, Overcoming the blood-brain tumor barrier for effective glioblastoma https://doi.org/10.1021/acs.nanolett.8b00390.
treatment, Drug Resist. Updat. 19 (2015) 1–12, https://doi.org/10.1016/j. [89] K. Mahmoudi, A. Bouras, D. Bozec, R. Ivkov, C. Hadjipanayis, Magnetic
drup.2015.02.002. hyperthermia therapy for the treatment of glioblastoma: a review of the therapy’s
[77] T.J. Lee, J.Y. Yoo, D. Shu, H. Li, J. Zhang, J.-G. Yu, A.C. Jaime-Ramirez, M. Acunzo, history, efficacy and application in humans, Int J Hyperthermia. 34 (2018)
G. Romano, R. Cui, H.-L. Sun, Z. Luo, M. Old, B. Kaur, P. Guo, C.M. Croce, RNA 1316–1328, https://doi.org/10.1080/02656736.2018.1430867.
Nanoparticle-Based Targeted Therapy for Glioblastoma through Inhibition of [90] J. Jose, R. Kumar, S. Harilal, G.E. Mathew, D.G.T. Parambi, A. Prabhu, Md.
Oncogenic miR-21, Mol Ther. 25 (2017) 1544–1555, https://doi.org/10.1016/j. S. Uddin, L. Aleya, H. Kim, B. Mathew, Magnetic nanoparticles for hyperthermia in
ymthe.2016.11.016. cancer treatment: an emerging tool, Environ Sci Pollut Res. (2019), https://doi.
[78] B. Küçüktürkmen, B. Devrim, O.M. Saka, Ş. Yilmaz, T. Arsoy, A. Bozkir, Co-delivery org/10.1007/s11356-019-07231-2.
of pemetrexed and miR-21 antisense oligonucleotide by lipid-polymer hybrid [91] M.S. Hasnain, Md.N. Javed, Md.S. Alam, P. Rishishwar, S. Rishishwar, S. Ali, A.
nanoparticles and effects on glioblastoma cells, Drug Dev Ind Pharm. 43 (2017) K. Nayak, S. Beg, Purple heart plant leaves extract-mediated silver nanoparticle
12–21, https://doi.org/10.1080/03639045.2016.1200069. synthesis: Optimization by Box-Behnken design, Materials Science and
[79] J.S. Ananta, R. Paulmurugan, T.F. Massoud, Nanoparticle-Delivered Antisense Engineering: C 99 (2019) 1105–1114, https://doi.org/10.1016/j.
MicroRNA-21 Enhances the Effects of Temozolomide on Glioblastoma Cells, Mol. msec.2019.02.061.
Pharmaceutics. 12 (2015) 4509–4517, https://doi.org/10.1021/acs. [92] E. Nduom, A. Bouras, M. Kaluzova, C.G. Hadjipanayis, Nanotechnology
molpharmaceut.5b00694. Applications for Glioblastoma, Neurosurg Clin N Am. 23 (2012) 439–449, https://
[80] P.M. Costa, A.L. Cardoso, C. Custódia, P. Cunha, L. Pereira de Almeida, M. doi.org/10.1016/j.nec.2012.04.006.
C. Pedroso de Lima, MiRNA-21 silencing mediated by tumor-targeted nanoparticles [93] N.Y. Hernández-Pedro, E. Rangel-López, R. Magaña-Maldonado, V.P. de la Cruz,
combined with sunitinib: A new multimodal gene therapy approach for A. Santamaría del Angel, B. Pineda, J. Sotelo, Application of Nanoparticles on
glioblastoma, J Control Release. 207 (2015) 31–39, https://doi.org/10.1016/j. Diagnosis and Therapy in Gliomas, Biomed Res Int. 2013 (2013), https://doi.org/
jconrel.2015.04.002. 10.1155/2013/351031.
[81] N.M. Teplyuk, E.J. Uhlmann, G. Gabriely, N. Volfovsky, Y. Wang, J. Teng, [94] U.K. Sukumar, R.J.C. Bose, M. Malhotra, H.A. Babikir, R. Afjei, E. Robinson,
P. Karmali, E. Marcusson, M. Peter, A. Mohan, Y. Kraytsberg, R. Cialic, E. Y. Zeng, E. Chang, F. Habte, R. Sinclair, S.S. Gambhir, T.F. Massoud,
A. Chiocca, J. Godlewski, B. Tannous, A.M. Krichevsky, Therapeutic potential of R. Paulmurugan, Intranasal delivery of targeted polyfunctional gold-iron oxide
targeting microRNA-10b in established intracranial glioblastoma: first steps toward nanoparticles loaded with therapeutic microRNAs for combined theranostic
the clinic, EMBO Molecular Medicine 8 (2016) 268–287, https://doi.org/ multimodality imaging and presensitization of glioblastoma to temozolomide,
10.15252/emmm.201505495. Biomaterials 218 (2019) 119342, https://doi.org/10.1016/j.
[82] Z. Shatsberg, X. Zhang, P. Ofek, S. Malhotra, A. Krivitsky, A. Scomparin, G. Tiram, biomaterials.2019.119342.
M. Calderón, R. Haag, R. Satchi-Fainaro, Functionalized nanogels carrying an [95] P.M. Costa, A.L. Cardoso, L.S. Mendonça, A. Serani, C. Custódia, M. Conceição,
anticancer microRNA for glioblastoma therapy, J Control Release. 239 (2016) S. Simões, J.N. Moreira, L. Pereira de Almeida, M.C. Pedroso de Lima, Tumor-
159–168, https://doi.org/10.1016/j.jconrel.2016.08.029. targeted Chlorotoxin-coupled Nanoparticles for Nucleic Acid Delivery to
[83] P. Ofek, M. Calderón, F.S. Mehrabadi, A. Krivitsky, S. Ferber, G. Tiram, Glioblastoma Cells: A Promising System for Glioblastoma Treatment, Mol Ther
N. Yerushalmi, S. Kredo-Russo, R. Grossman, Z. Ram, R. Haag, R. Satchi-Fainaro, Nucleic Acids. 2 (2013) e100, https://doi.org/10.1038/mtna.2013.30.
Restoring the oncosuppressor activity of microRNA-34a in glioblastoma using a [96] Y.-E. Seo, H.-W. Suh, R. Bahal, A. Josowitz, J. Zhang, E. Song, J. Cui,
polyglycerol-based polyplex, Nanomedicine: Nanotechnology, Biology and S. Noorbakhsh, C. Jackson, T. Bu, A. Piotrowski-Daspit, R. Bindra, W.M. Saltzman,
Medicine. 12 (2016) 2201–2214, https://doi.org/10.1016/j.nano.2016.05.016. Nanoparticle-mediated intratumoral inhibition of miR-21 for improved survival in
[84] G. Jiang, H. Chen, J. Huang, Q. Song, Y. Chen, X. Gu, Z. Jiang, Y. Huang, Y. Lin, glioblastoma, Biomaterials 201 (2019) 87–98, https://doi.org/10.1016/j.
J. Feng, J. Jiang, Y. Bao, G. Zheng, J. Chen, H. Chen, X. Gao, Tailored Lipoprotein- biomaterials.2019.02.016.

398

Downloaded for Anonymous User (n/a) at Autonomous University of Guadalajara from ClinicalKey.com by Elsevier on February
28, 2023. For personal use only. No other uses without permission. Copyright ©2023. Elsevier Inc. All rights reserved.

You might also like